#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Analysis of the SUMO2 Proteome during HSV-1 Infection


Proteins are subject to many types of modification that regulate their functions and which are applied after their initial synthesis in the cell. One such modification is known as sumoylation, the covalent linkage of a small ubiquitin-like protein to a wide variety of substrate proteins. Sumoylation is involved in the regulation of many cellular pathways, including transcription, DNA repair, chromatin modification and defence to viral infections. Several viruses have connections with sumoylation, either through modification of their own proteins or in changing the sumoylation status of cellular proteins in ways that may be beneficial for infection. Herpes simplex virus type 1 (HSV-1) causes a widespread reduction in uncharacterized sumoylated cellular protein species, an effect that is caused by one of its key regulatory proteins (ICP0), which also induces the degradation of a number of repressive cellular proteins and thereby stimulates efficient infection. This study describes a comprehensive analysis of cellular proteins whose sumoylation status is altered by HSV-1 infection. Of 877 putative cellular sumoylation substrates, we found 124 whose sumoylation status reduces at least three-fold during infection. We validated the behavior of several such proteins and identified amongst them several novel targets of ICP0 activity with predicted repressive properties.


Vyšlo v časopise: Analysis of the SUMO2 Proteome during HSV-1 Infection. PLoS Pathog 11(7): e32767. doi:10.1371/journal.ppat.1005059
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1005059

Souhrn

Proteins are subject to many types of modification that regulate their functions and which are applied after their initial synthesis in the cell. One such modification is known as sumoylation, the covalent linkage of a small ubiquitin-like protein to a wide variety of substrate proteins. Sumoylation is involved in the regulation of many cellular pathways, including transcription, DNA repair, chromatin modification and defence to viral infections. Several viruses have connections with sumoylation, either through modification of their own proteins or in changing the sumoylation status of cellular proteins in ways that may be beneficial for infection. Herpes simplex virus type 1 (HSV-1) causes a widespread reduction in uncharacterized sumoylated cellular protein species, an effect that is caused by one of its key regulatory proteins (ICP0), which also induces the degradation of a number of repressive cellular proteins and thereby stimulates efficient infection. This study describes a comprehensive analysis of cellular proteins whose sumoylation status is altered by HSV-1 infection. Of 877 putative cellular sumoylation substrates, we found 124 whose sumoylation status reduces at least three-fold during infection. We validated the behavior of several such proteins and identified amongst them several novel targets of ICP0 activity with predicted repressive properties.


Zdroje

1. Knipe DM, Howley PM, Griffin DE, Lamb RA, Martin MA, et al. (2006) Fields Virology. Philadelphia: Lippincott Williams and Wilkins.

2. Weller SK (2011) Alphaherpesviruses. Molecular Virology. Norfolk, U.K.: Caister Academic Press.

3. Randall RE, Goodbourn S (2008) Interferons and viruses: an interplay between induction, signalling, antiviral responses and virus countermeasures. J Gen Virol 89: 1–47. 18089727

4. Tavalai N, Stamminger T (2008) New insights into the role of the subnuclear structure ND10 for viral infection. Biochim Biophys Acta 1783: 2207–2221. doi: 10.1016/j.bbamcr.2008.08.004 18775455

5. Sternsdorf T, Jensen K, Will H (1997) Evidence for covalent modification of the nuclear dot-associated proteins PML and Sp100 by PIC1/SUMO-1. J Cell Biol 139: 1621–1634. 9412458

6. Bernardi R, Pandolfi PP (2007) Structure, dynamics and functions of promyelocytic leukaemia nuclear bodies. Nat Rev Mol Cell Biol 8: 1006–1016. 17928811

7. Shen TH, Lin HK, Scaglioni PP, Yung TM, Pandolfi PP (2006) The mechanisms of PML-nuclear body formation. Mol Cell 24: 331–339. 17081985

8. Everett RD, Murray J (2005) ND10 Components Relocate to Sites Associated with Herpes Simplex Virus Type 1 Nucleoprotein Complexes during Virus Infection. J Virol 79: 5078–5089. 15795293

9. Everett RD, Parada C, Gripon P, Sirma H, Orr A (2008) Replication of ICP0-null mutant herpes simplex virus type 1 is restricted by both PML and Sp100. J Virol 82: 2661–2672. 18160441

10. Everett RD, Rechter S, Papior P, Tavalai N, Stamminger T, et al. (2006) PML contributes to a cellular mechanism of repression of herpes simplex virus type 1 infection that is inactivated by ICP0. J Virol 80: 7995–8005. 16873256

11. Glass M, Everett RD (2013) Components of promyelocytic leukemia nuclear bodies (ND10) act cooperatively to repress herpesvirus infection. J Virol 87: 2174–2185. doi: 10.1128/JVI.02950-12 23221561

12. Lukashchuk V, Orr A, Everett RD (2010) Regulation of ICP0 null mutant HSV-1 infection by ND10 components ATRX and hDaxx. J Virol 84: 4026–4040. doi: 10.1128/JVI.02597-09 20147399

13. Cuchet-Lourenco D, Boutell C, Lukashchuk V, Grant K, Sykes A, et al. (2011) SUMO Pathway Dependent Recruitment of Cellular Repressors to Herpes Simplex Virus Type 1 Genomes. PLoS Pathog 7: e1002123. doi: 10.1371/journal.ppat.1002123 21779164

14. Eidson KM, Hobbs WE, Manning BJ, Carlson P, DeLuca NA (2002) Expression of herpes simplex virus ICP0 inhibits the induction of interferon-stimulated genes by viral infection. J Virol 76: 2180–2191. 11836395

15. Harle P, Sainz B Jr., Carr DJ, Halford WP (2002) The immediate-early protein, ICP0, is essential for the resistance of herpes simplex virus to interferon-alpha/beta. Virology 293: 295–304. 11886249

16. Mossman KL, Saffran HA, Smiley JR (2000) Herpes simplex virus ICP0 mutants are hypersensitive to interferon. J Virol 74: 2052–2056. 10644380

17. Boutell C, Everett RD (2013) Regulation of alphaherpesvirus infections by the ICP0 family of proteins. J Gen Virol 94: 465–481. doi: 10.1099/vir.0.048900-0 23239572

18. Boutell C, Cuchet-Lourenco D, Vanni E, Orr A, Glass M, et al. (2011) A Viral Ubiquitin Ligase Has Substrate Preferential SUMO Targeted Ubiquitin Ligase Activity that Counteracts Intrinsic Antiviral Defence. PLoS Pathog 7: e1002245. doi: 10.1371/journal.ppat.1002245 21949651

19. Everett RD, Freemont P, Saitoh H, Dasso M, Orr A, et al. (1998) The disruption of ND10 during herpes simplex virus infection correlates with the Vmw110- and proteasome-dependent loss of several PML isoforms. J Virol 72: 6581–6591. 9658103

20. Cuchet-Lourenco D, Vanni E, Glass M, Orr A, Everett RD (2012) Herpes Simplex Virus 1 Ubiquitin Ligase ICP0 Interacts with PML Isoform I and Induces Its SUMO-Independent Degradation. J Virol 86: 11209–11222. 22875967

21. Gareau JR, Lima CD (2010) The SUMO pathway: emerging mechanisms that shape specificity, conjugation and recognition. Nat Rev Mol Cell Biol 11: 861–871. doi: 10.1038/nrm3011 21102611

22. Wang Y, Dasso M (2009) SUMOylation and deSUMOylation at a glance. J Cell Sci 122: 4249–4252. doi: 10.1242/jcs.050542 19923268

23. Becker J, Barysch SV, Karaca S, Dittner C, Hsiao HH, et al. (2013) Detecting endogenous SUMO targets in mammalian cells and tissues. Nat Struct Mol Biol 20: 525–531. doi: 10.1038/nsmb.2526 23503365

24. Golebiowski F, Matic I, Tatham MH, Cole C, Yin Y, et al. (2009) System-wide changes to SUMO modifications in response to heat shock. Sci Signal 2: ra24. doi: 10.1126/scisignal.2000282 19471022

25. Hendriks IA, D'Souza RC, Yang B, Verlaan-de Vries M, Mann M, et al. (2014) Uncovering global SUMOylation signaling networks in a site-specific manner. Nat Struct Mol Biol 21: 927–936. doi: 10.1038/nsmb.2890 25218447

26. Tammsalu T, Matic I, Jaffray EG, Ibrahim AF, Tatham MH, et al. (2014) Proteome-Wide Identification of SUMO2 Modification Sites. Sci Signal 7: rs2. doi: 10.1126/scisignal.2005146 24782567

27. Tatham MH, Rodriguez MS, Xirodimas DP, Hay RT (2009) Detection of protein SUMOylation in vivo. Nat Protoc 4: 1363–1371. doi: 10.1038/nprot.2009.128 19730420

28. Impens F, Radoshevich L, Cossart P, Ribet D (2014) Mapping of SUMO sites and analysis of SUMOylation changes induced by external stimuli. Proc Natl Acad Sci U S A 111: 12432–12437. doi: 10.1073/pnas.1413825111 25114211

29. Tatham MH, Matic I, Mann M, Hay RT (2011) Comparative proteomic analysis identifies a role for SUMO in protein quality control. Sci Signal 4: rs4. doi: 10.1126/scisignal.2001484 21693764

30. Cox J, Mann M (2008) MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat Biotechnol 26: 1367–1372. doi: 10.1038/nbt.1511 19029910

31. Cuchet-Lourenco D, Anderson G, Sloan E, Orr A, Everett RD (2013) The viral ubiquitin ligase ICP0 is neither sufficient nor necessary for degradation of the cellular DNA sensor IFI16 during herpes simplex virus 1 infection. J Virol 87: 13422–13432. doi: 10.1128/JVI.02474-13 24089555

32. Orzalli MH, DeLuca NA, Knipe DM (2012) Nuclear IFI16 induction of IRF-3 signaling during herpesviral infection and degradation of IFI16 by the viral ICP0 protein. Proc Natl Acad Sci U S A 109: E3008–3017. doi: 10.1073/pnas.1211302109 23027953

33. Lomonte P, Morency E (2007) Centromeric protein CENP-B proteasomal degradation induced by the viral protein ICP0. FEBS Lett 581: 658–662. 17258208

34. Kwong AD, Frenkel N (1989) The herpes simplex virus virion host shutoff function. J Virol 63: 4834–4839. 2552156

35. Davison MD, Rixon FJ, Davison AJ (1992) Identification of genes encoding two capsid proteins (VP24 and VP26) of herpes simplex virus type 1. J Gen Virol 73 (Pt 10): 2709–2713. 1328483

36. Liu FY, Roizman B (1991) The promoter, transcriptional unit, and coding sequence of herpes simplex virus 1 family 35 proteins are contained within and in frame with the UL26 open reading frame. J Virol 65: 206–212. 1845885

37. Sinigalia E, Alvisi G, Segre CV, Mercorelli B, Muratore G, et al. (2012) The human cytomegalovirus DNA polymerase processivity factor UL44 is modified by SUMO in a DNA-dependent manner. PLoS One 7: e49630. doi: 10.1371/journal.pone.0049630 23166733

38. Everett RD, Earnshaw WC, Findlay J, Lomonte P (1999) Specific destruction of kinetochore protein CENP-C and disruption of cell division by herpes simplex virus immediate-early protein Vmw110. EMBO J 18: 1526–1538. 10075924

39. Lomonte P, Sullivan KF, Everett RD (2001) Degradation of nucleosome-associated centromeric histone H3-like protein CENP-A induced by herpes simplex virus type 1 protein ICP0. J Biol Chem 276: 5829–5835. 11053442

40. Muller S, Dejean A (1999) Viral immediate-early proteins abrogate the modification by SUMO-1 of PML and Sp100 proteins, correlating with nuclear body disruption. J Virol 73: 5137–5143. 10233977

41. Parkinson J, Everett RD (2000) Alphaherpesvirus proteins related to herpes simplex virus type 1 ICP0 affect cellular structures and proteins. J Virol 74: 10006–10017. 11024129

42. Everett RD, Parsy ML, Orr A (2009) Analysis of the functions of herpes simplex virus type 1 regulatory protein ICP0 that are critical for lytic infection and derepression of quiescent viral genomes. J Virol 83: 4963–4977. doi: 10.1128/JVI.02593-08 19264778

43. Fukuyo Y, Horikoshi N, Ishov AM, Silverstein SJ, Nakajima T (2011) The herpes simplex virus immediate-early ubiquitin ligase ICP0 induces degradation of the ICP0 repressor protein E2FBP1. J Virol 85: 3356–3366. doi: 10.1128/JVI.02105-10 21248039

44. Everett RD, Boutell C, Pheasant K, Cuchet-Lourenco D, Orr A (2014) Sequences related to SUMO interaction motifs in herpes simplex virus 1 protein ICP0 act cooperatively to stimulate virus infection. J Virol 88: 2763–2774. doi: 10.1128/JVI.03417-13 24352468

45. Chelbi-Alix MK, de The H (1999) Herpes virus induced proteasome-dependent degradation of the nuclear bodies-associated PML and Sp100 proteins. Oncogene 18: 935–941. 10023669

46. Liu B, Shuai K (2008) Regulation of the sumoylation system in gene expression. Curr Opin Cell Biol 20: 288–293. doi: 10.1016/j.ceb.2008.03.014 18468876

47. Gill G (2005) Something about SUMO inhibits transcription. Curr Opin Genet Dev 15: 536–541. 16095902

48. Hay RT (2005) SUMO: a history of modification. Mol Cell 18: 1–12. 15808504

49. Lyst MJ, Stancheva I (2007) A role for SUMO modification in transcriptional repression and activation. Biochem Soc Trans 35: 1389–1392. 18031228

50. Gill G (2003) Post-translational modification by the small ubiquitin-related modifier SUMO has big effects on transcription factor activity. Curr Opin Genet Dev 13: 108–113. 12672486

51. Verger A, Perdomo J, Crossley M (2003) Modification with SUMO. A role in transcriptional regulation. EMBO Rep 4: 137–142. 12612601

52. Hay RT (2006) Role of ubiquitin-like proteins in transcriptional regulation. Ernst Schering Res Found Workshop: 173–192. 16568955

53. Kotaja N, Karvonen U, Janne OA, Palvimo JJ (2002) The nuclear receptor interaction domain of GRIP1 is modulated by covalent attachment of SUMO-1. J Biol Chem 277: 30283–30288. 12060666

54. Gill G (2004) SUMO and ubiquitin in the nucleus: different functions, similar mechanisms? Genes Dev 18: 2046–2059. 15342487

55. Psakhye I, Jentsch S (2012) Protein group modification and synergy in the SUMO pathway as exemplified in DNA repair. Cell 151: 807–820. doi: 10.1016/j.cell.2012.10.021 23122649

56. Siggs OM, Beutler B (2012) The BTB-ZF transcription factors. Cell Cycle 11: 3358–3369. doi: 10.4161/cc.21277 22894929

57. Perez-Torrado R, Yamada D, Defossez PA (2006) Born to bind: the BTB protein-protein interaction domain. Bioessays 28: 1194–1202. 17120193

58. Sathyan KM, Shen Z, Tripathi V, Prasanth KV, Prasanth SG (2011) A BEN-domain-containing protein associates with heterochromatin and represses transcription. J Cell Sci 124: 3149–3163. doi: 10.1242/jcs.086603 21914818

59. Takahashi K, Yoshida N, Murakami N, Kawata K, Ishizaki H, et al. (2007) Dynamic regulation of p53 subnuclear localization and senescence by MORC3. Mol Biol Cell 18: 1701–1709. 17332504

60. Mimura Y, Takahashi K, Kawata K, Akazawa T, Inoue N (2010) Two-step colocalization of MORC3 with PML nuclear bodies. J Cell Sci 123: 2014–2024. doi: 10.1242/jcs.063586 20501696

61. Clouaire T, de Las Heras JI, Merusi C, Stancheva I (2010) Recruitment of MBD1 to target genes requires sequence-specific interaction of the MBD domain with methylated DNA. Nucleic Acids Res 38: 4620–4634. doi: 10.1093/nar/gkq228 20378711

62. Klose RJ, Bird AP (2006) Genomic DNA methylation: the mark and its mediators. Trends Biochem Sci 31: 89–97. 16403636

63. Ng HH, Jeppesen P, Bird A (2000) Active repression of methylated genes by the chromosomal protein MBD1. Mol Cell Biol 20: 1394–1406. 10648624

64. Fujita N, Shimotake N, Ohki I, Chiba T, Saya H, et al. (2000) Mechanism of transcriptional regulation by methyl-CpG binding protein MBD1. Mol Cell Biol 20: 5107–5118. 10866667

65. Chakrabarti SR, Sood R, Nandi S, Nucifora G (2000) Posttranslational modification of TEL and TEL/AML1 by SUMO-1 and cell-cycle-dependent assembly into nuclear bodies. Proc Natl Acad Sci U S A 97: 13281–13285. 11078523

66. Chakrabarti SR, Sood R, Ganguly S, Bohlander S, Shen Z, et al. (1999) Modulation of TEL transcription activity by interaction with the ubiquitin-conjugating enzyme UBC9. Proc Natl Acad Sci U S A 96: 7467–7472. 10377438

67. Chakrabarti SR, Nucifora G (1999) The leukemia-associated gene TEL encodes a transcription repressor which associates with SMRT and mSin3A. Biochem Biophys Res Commun 264: 871–877. 10544023

68. Guidez F, Petrie K, Ford AM, Lu H, Bennett CA, et al. (2000) Recruitment of the nuclear receptor corepressor N-CoR by the TEL moiety of the childhood leukemia-associated TEL-AML1 oncoprotein. Blood 96: 2557–2561. 11001911

69. Wang L, Hiebert SW (2001) TEL contacts multiple co-repressors and specifically associates with histone deacetylase-3. Oncogene 20: 3716–3725. 11439334

70. Fenrick R, Amann JM, Lutterbach B, Wang L, Westendorf JJ, et al. (1999) Both TEL and AML-1 contribute repression domains to the t(12;21) fusion protein. Mol Cell Biol 19: 6566–6574. 10490596

71. Jousset C, Carron C, Boureux A, Quang CT, Oury C, et al. (1997) A domain of TEL conserved in a subset of ETS proteins defines a specific oligomerization interface essential to the mitogenic properties of the TEL-PDGFR beta oncoprotein. EMBO J 16: 69–82. 9009269

72. Orzalli MH, Conwell SE, Berrios C, DeCaprio JA, Knipe DM (2013) Nuclear interferon-inducible protein 16 promotes silencing of herpesviral and transfected DNA. Proc Natl Acad Sci U S A 110: E4492–4501. doi: 10.1073/pnas.1316194110 24198334

73. Adamson AL (2005) Effects of SUMO-1 upon Epstein-Barr virus BZLF1 function and BMRF1 expression. Biochem Biophys Res Commun 336: 22–28. 16112644

74. Everett RD, Boutell C, Hale BG (2013) Interplay between viruses and host sumoylation pathways. Nat Rev Microbiol 11: 400–411. doi: 10.1038/nrmicro3015 23624814

75. Wimmer P, Schreiner S, Dobner T (2012) Human pathogens and the host cell SUMOylation system. J Virol 86: 642–654. doi: 10.1128/JVI.06227-11 22072786

76. Gripon P, Rumin S, Urban S, Le Seyec J, Glaise D, et al. (2002) Infection of a human hepatoma cell line by hepatitis B virus. Proc Natl Acad Sci U S A 99: 15655–15660. 12432097

77. Stow ND, Stow EC (1986) Isolation and characterization of a herpes simplex virus type 1 mutant containing a deletion within the gene encoding the immediate early polypeptide Vmw110. J Gen Virol 67: 2571–2585. 3025339

78. Yao F, Schaffer PA (1995) An activity specified by the osteosarcoma line U2OS can substitute functionally for ICP0, a major regulatory protein of herpes simplex virus type 1. J Virol 69: 6249–6258. 7666525

79. Cuchet D, Sykes A, Nicolas A, Orr A, Murray J, et al. (2011) PML isoforms I and II participate in PML-dependent restriction of HSV-1 replication. J Cell Sci 124: 280–291. doi: 10.1242/jcs.075390 21172801

80. Shevchenko A, Tomas H, Havlis J, Olsen J, Mann M (2007) In-gel digestion for mass spectrometric characterization of proteins and proteomes. Nature Protocols 1: 2856–2860.

81. Cox J, Neuhauser N, Michalski A, Scheltema RA, Olsen JV, et al. (2011) Andromeda: a peptide search engine integrated into the MaxQuant environment. J Proteome Res 10: 1794–1805. doi: 10.1021/pr101065j 21254760

82. Stuurman N, de Graaf A, Floore A, Josso A, Humbel B, et al. (1992) A monoclonal antibody recognizing nuclear matrix-associated nuclear bodies. J Cell Sci 101: 773–784. 1527179

83. Filion GJ, Zhenilo S, Salozhin S, Yamada D, Prokhortchouk E, et al. (2006) A family of human zinc finger proteins that bind methylated DNA and repress transcription. Mol Cell Biol 26: 169–181. 16354688

84. Kim K, Chadalapaka G, Pathi SS, Jin UH, Lee JS, et al. (2012) Induction of the transcriptional repressor ZBTB4 in prostate cancer cells by drug-induced targeting of microRNA-17-92/106b-25 clusters. Mol Cancer Ther 11: 1852–1862. doi: 10.1158/1535-7163.MCT-12-0181 22752225

85. Weber A, Marquardt J, Elzi D, Forster N, Starke S, et al. (2008) Zbtb4 represses transcription of P21CIP1 and controls the cellular response to p53 activation. EMBO J 27: 1563–1574. doi: 10.1038/emboj.2008.85 18451802

86. Tillotson LG (1999) RIN ZF, a novel zinc finger gene, encodes proteins that bind to the CACC element of the gastrin promoter. J Biol Chem 274: 8123–8128. 10075714

87. Mertens-Talcott SU, Chintharlapalli S, Li X, Safe S (2007) The oncogenic microRNA-27a targets genes that regulate specificity protein transcription factors and the G2-M checkpoint in MDA-MB-231 breast cancer cells. Cancer Res 67: 11001–11011. 18006846

88. Pan Q, Ning Y, Chen LZ, Zhang S, Liu ZZ, et al. (2012) Association of MHC class-III gene polymorphisms with ER-positive breast cancer in Chinese Han population. Genet Mol Res 11: 4299–4306. doi: 10.4238/2012.September.17.1 23079975

89. Tirard M, Hsiao HH, Nikolov M, Urlaub H, Melchior F, et al. (2012) In vivo localization and identification of SUMOylated proteins in the brain of His6-HA-SUMO1 knock-in mice. Proc Natl Acad Sci U S A 109: 21122–21127. doi: 10.1073/pnas.1215366110 23213215

90. Zhang W, Mi J, Li N, Sui L, Wan T, et al. (2001) Identification and characterization of DPZF, a novel human BTB/POZ zinc finger protein sharing homology to BCL-6. Biochem Biophys Res Commun 282: 1067–1073. 11352661

91. Liu X, Zhang P, Bao Y, Han Y, Wang Y, et al. (2013) Zinc finger protein ZBTB20 promotes toll-like receptor-triggered innate immune responses by repressing IkappaBalpha gene transcription. Proc Natl Acad Sci U S A.

92. Sasai N, Matsuda E, Sarashina E, Ishida Y, Kawaichi M (2005) Identification of a novel BTB-zinc finger transcriptional repressor, CIBZ, that interacts with CtBP corepressor. Genes Cells 10: 871–885. 16115196

93. Abhiman S, Iyer LM, Aravind L (2008) BEN: a novel domain in chromatin factors and DNA viral proteins. Bioinformatics 24: 458–461. doi: 10.1093/bioinformatics/btn007 18203771

94. Korutla L, Degnan R, Wang P, Mackler SA (2007) NAC1, a cocaine-regulated POZ/BTB protein interacts with CoREST. J Neurochem 101: 611–618. 17254023

95. Korutla L, Wang PJ, Mackler SA (2005) The POZ/BTB protein NAC1 interacts with two different histone deacetylases in neuronal-like cultures. J Neurochem 94: 786–793. 16033423

96. Shen H, Korutla L, Champtiaux N, Toda S, LaLumiere R, et al. (2007) NAC1 regulates the recruitment of the proteasome complex into dendritic spines. J Neurosci 27: 8903–8913. 17699672

97. Nakayama K, Rahman MT, Rahman M, Yeasmin S, Ishikawa M, et al. (2010) Biological role and prognostic significance of NAC1 in ovarian cancer. Gynecol Oncol 119: 469–478. doi: 10.1016/j.ygyno.2010.08.031 20869761

98. Xuan C, Wang Q, Han X, Duan Y, Li L, et al. (2013) RBB, a novel transcription repressor, represses the transcription of HDM2 oncogene. Oncogene 32: 3711–3721. doi: 10.1038/onc.2012.386 22926524

99. van den Berg DL, Snoek T, Mullin NP, Yates A, Bezstarosti K, et al. (2010) An Oct4-centered protein interaction network in embryonic stem cells. Cell Stem Cell 6: 369–381. doi: 10.1016/j.stem.2010.02.014 20362541

100. Prieur A, Nacerddine K, van Lohuizen M, Peeper DS (2009) SUMOylation of DRIL1 directs its transcriptional activity towards leukocyte lineage-specific genes. PLoS One 4: e5542. doi: 10.1371/journal.pone.0005542 19436740

101. Ma K, Araki K, Ichwan SJ, Suganuma T, Tamamori-Adachi M, et al. (2003) E2FBP1/DRIL1, an AT-rich interaction domain-family transcription factor, is regulated by p53. Mol Cancer Res 1: 438–444. 12692263

102. Suzuki M, Okuyama S, Okamoto S, Shirasuna K, Nakajima T, et al. (1998) A novel E2F binding protein with Myc-type HLH motif stimulates E2F-dependent transcription by forming a heterodimer. Oncogene 17: 853–865. 9780002

103. Fukuyo Y, Mogi K, Tsunematsu Y, Nakajima T (2004) E2FBP1/hDril1 modulates cell growth through downregulation of promyelocytic leukemia bodies. Cell Death Differ 11: 747–759. 15017387

104. Fiorentino DF, Chung LS, Christopher-Stine L, Zaba L, Li S, et al. (2013) Most patients with cancer-associated dermatomyositis have antibodies to nuclear matrix protein NXP-2 or transcription intermediary factor 1gamma. Arthritis Rheum 65: 2954–2962. doi: 10.1002/art.38093 24037894

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2015 Číslo 7
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#